Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 6909, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37907525

ABSTRACT

Osteoarthritis (OA) is characterised by an irreversible degeneration of articular cartilage. Here we show that the BMP-antagonist Gremlin 1 (Grem1) marks a bipotent chondrogenic and osteogenic progenitor cell population within the articular surface. Notably, these progenitors are depleted by injury-induced OA and increasing age. OA is also caused by ablation of Grem1 cells in mice. Transcriptomic and functional analysis in mice found that articular surface Grem1-lineage cells are dependent on Foxo1 and ablation of Foxo1 in Grem1-lineage cells caused OA. FGFR3 signalling was confirmed as a promising therapeutic pathway by administration of pathway activator, FGF18, resulting in Grem1-lineage chondrocyte progenitor cell proliferation, increased cartilage thickness and reduced OA. These findings suggest that OA, in part, is caused by mechanical, developmental or age-related attrition of Grem1 expressing articular cartilage progenitor cells. These cells, and the FGFR3 signalling pathway that sustains them, may be effective future targets for biological management of OA.


Subject(s)
Cartilage, Articular , Osteoarthritis , Mice , Animals , Osteoarthritis/genetics , Osteoarthritis/metabolism , Stem Cells/metabolism , Cells, Cultured , Gene Expression Profiling , Osteogenesis , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Intercellular Signaling Peptides and Proteins/metabolism
2.
bioRxiv ; 2023 Mar 30.
Article in English | MEDLINE | ID: mdl-37034712

ABSTRACT

Osteoarthritis (OA), which carries an enormous disease burden across the world, is characterised by irreversible degeneration of articular cartilage (AC), and subsequently bone. The cellular cause of OA is unknown. Here, using lineage tracing in mice, we show that the BMP-antagonist Gremlin 1 (Grem1) marks a novel chondrogenic progenitor (CP) cell population in the articular surface that generates joint cartilage and subchondral bone during development and adulthood. Notably, this CP population is depleted in injury-induced OA, and with age. OA is also induced by toxin-mediated ablation of Grem1 CP cells in young mice. Transcriptomic analysis and functional modelling in mice revealed articular surface Grem1-lineage cells are dependent on Foxo1; ablation of Foxo1 in Grem1-lineage cells led to early OA. This analysis identified FGFR3 signalling as a therapeutic target, and injection of its activator, FGF18, caused proliferation of Grem1-lineage CP cells, increased cartilage thickness, and reduced OA pathology. We propose that OA arises from the loss of CP cells at the articular surface secondary to an imbalance in progenitor cell homeostasis and present a new progenitor population as a locus for OA therapy.

3.
Int J Mol Sci ; 23(2)2022 Jan 15.
Article in English | MEDLINE | ID: mdl-35055125

ABSTRACT

PTH induces phosphorylation of the transcriptional coregulator NACA on serine 99 through Gαs and PKA. This leads to nuclear translocation of NACA and expression of the target gene Lrp6, encoding a coreceptor of the PTH receptor (PTH1R) necessary for full anabolic response to intermittent PTH (iPTH) treatment. We hypothesized that maintaining enough functional PTH1R/LRP6 coreceptor complexes at the plasma membrane through NACA-dependent Lrp6 transcription is important to ensure maximal response to iPTH. To test this model, we generated compound heterozygous mice in which one allele each of Naca and Lrp6 is inactivated in osteoblasts and osteocytes, using a knock-in strain with a Naca99 Ser-to-Ala mutation and an Lrp6 floxed strain (test genotype: Naca99S/A; Lrp6+/fl;OCN-Cre). Four-month-old females were injected with vehicle or 100 µg/kg PTH(1-34) once daily, 5 days a week for 4 weeks. Control mice showed significant increases in vertebral trabecular bone mass and biomechanical properties that were abolished in compound heterozygotes. Lrp6 expression was reduced in compound heterozygotes vs. controls. The iPTH treatment increased Alpl and Col1a1 mRNA levels in the control but not in the test group. These results confirm that NACA and LRP6 form part of a common genetic pathway that is necessary for the full anabolic effect of iPTH.


Subject(s)
Anabolic Agents/administration & dosage , Embryonic Stem Cells/cytology , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Molecular Chaperones/genetics , Parathyroid Hormone/administration & dosage , Anabolic Agents/pharmacology , Animals , Cell Line , Cell Membrane/metabolism , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , Gene Knock-In Techniques , Mice , Molecular Chaperones/metabolism , Mutagenesis, Site-Directed , Osteoblasts/metabolism , Osteocytes/metabolism , Parathyroid Hormone/pharmacology , Phosphorylation , Signal Transduction/drug effects , X-Ray Microtomography
4.
Proc Natl Acad Sci U S A ; 116(24): 11978-11987, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31138698

ABSTRACT

Antigen-directed immunotherapies for acute myeloid leukemia (AML), such as chimeric antigen receptor T cells (CAR-Ts) or antibody-drug conjugates (ADCs), are associated with severe toxicities due to the lack of unique targetable antigens that can distinguish leukemic cells from normal myeloid cells or myeloid progenitors. Here, we present an approach to treat AML by targeting the lineage-specific myeloid antigen CD33. Our approach combines CD33-targeted CAR-T cells, or the ADC Gemtuzumab Ozogamicin with the transplantation of hematopoietic stem cells that have been engineered to ablate CD33 expression using genomic engineering methods. We show highly efficient genetic ablation of CD33 antigen using CRISPR/Cas9 technology in human stem/progenitor cells (HSPC) and provide evidence that the deletion of CD33 in HSPC doesn't impair their ability to engraft and to repopulate a functional multilineage hematopoietic system in vivo. Whole-genome sequencing and RNA sequencing analysis revealed no detectable off-target mutagenesis and no loss of functional p53 pathways. Using a human AML cell line (HL-60), we modeled a postremission marrow with minimal residual disease and showed that the transplantation of CD33-ablated HSPCs with CD33-targeted immunotherapy leads to leukemia clearance, without myelosuppression, as demonstrated by the engraftment and recovery of multilineage descendants of CD33-ablated HSPCs. Our study thus contributes to the advancement of targeted immunotherapy and could be replicated in other malignancies.

5.
J Cell Biochem ; 115(5): 866-73, 2014 May.
Article in English | MEDLINE | ID: mdl-24375853

ABSTRACT

The transcriptional coregulator αNAC (Nascent polypeptide associated complex And Coregulator alpha) and the transcriptional repressor FIAT (Factor Inhibiting ATF4-mediated Transcription) interact but the biological relevance of this interaction remains unclear. The activity of αNAC is extensively modulated by post-translational modifications (PTMs). We identified a novel αNAC PTM through covalent attachment of the Small Ubiquitin-like MOdifier (SUMO1). Recombinant αNAC was a SUMO1 target in in vitro SUMOylation assays and we confirmed that αNAC is conjugated to SUMO1 in cultured osteoblasts and in calvarial tissue. The amino acid sequence of αNAC contains one copy of the composite "phospho-sumoyl switch" motif that couples sequential phosphorylation and SUMOylation. We found that αNAC is selectively SUMOylated at lysine residue 127 within the motif and that SUMOylation is enhanced when a phosphomimetic mutation is introduced at the nearby serine residue 132. SUMOylation did not alter the DNA-binding capacity of αNAC. The S132D, hyper-SUMOylated αNAC mutant specifically interacted with histone deacetylase-2 (HDAC2) and enhanced the inhibitory activity of FIAT on ATF4-mediated transcription from the Osteocalcin gene promoter. This effect required binding of SUMOylated αNAC to the target promoter. We propose that maximal transcriptional repression by FIAT requires its interaction with SUMOylated, HDAC2-interacting αNAC.


Subject(s)
Co-Repressor Proteins/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Sumoylation/genetics , Transcription, Genetic , Animals , Co-Repressor Proteins/genetics , Histone Deacetylase 2/metabolism , Mice , Molecular Chaperones/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/genetics , Phosphorylation/genetics , Promoter Regions, Genetic , Protein Processing, Post-Translational/genetics
6.
Biochim Biophys Acta ; 1819(11-12): 1208-16, 2012.
Article in English | MEDLINE | ID: mdl-23092676

ABSTRACT

In the nucleus of differentiated osteoblasts, the DNA-binding αNAC protein acts as a transcriptional coactivator of the Osteocalcin gene. Chromatin immunoprecipitation-microarray assays (ChIP-chip) showed that αNAC binds the Osteocalcin promoter but also identified the Myogenin promoter as an αNAC target. Here, we confirm these array data using quantitative ChIP and further detected that αNAC binds to these promoters in myoblasts. Since these genes are differentially regulated during osteoblastogenesis or myogenesis, these results suggest cell- and promoter-context specific functions for αNAC. We hypothesized that αNAC dynamically recruits corepressors to inhibit Myogenin expression in cells committing to the osteoblastic lineage or to inhibit Osteocalcin transcription in differentiating myoblasts. Using co-immunoprecipitation assays, we detected complexes between αNAC and the corepressors HDAC1 and HDAC3, in myoblasts and osteoblasts. Sequential ChIP confirmed HDAC1 recruitment by αNAC at the Osteocalcin and Myogenin promoters. Interaction with the corepressors was detectable in pre-osteoblasts and in myoblasts but disappeared as the cells differentiate. Treatment with an HDAC inhibitor caused de-repression of Osteocalcin expression in myoblasts. Overexpression of αNAC in myoblasts inhibits expression of Myogenin and differentiation. However, overexpression of an N-terminus truncated αNAC mutant allowed myoblasts to express Myogenin and differentiate, and this mutant did not interact with HDAC1 or HDAC3. This study identified an additional DNA-binding target and novel protein-protein interactions for αNAC. We propose that αNAC plays a role in regulating gene transcription during mesenchymal cell differentiation by differentially recruiting corepressors at target promoters.


Subject(s)
Gene Expression Regulation/physiology , Histone Deacetylase 1/metabolism , Histone Deacetylases/metabolism , Molecular Chaperones/metabolism , Myoblasts/metabolism , Myogenin/biosynthesis , Osteoblasts/metabolism , Osteocalcin/biosynthesis , Promoter Regions, Genetic/physiology , Animals , Cell Differentiation/physiology , Cell Line , Histone Deacetylase 1/genetics , Histone Deacetylases/genetics , Mice , Molecular Chaperones/genetics , Myoblasts/cytology , Myogenin/genetics , Osteoblasts/cytology , Osteocalcin/genetics , Transcription, Genetic/physiology
7.
Am J Orthod Dentofacial Orthop ; 138(2): 140.e1-140.e11; discussion 140-1, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20691350

ABSTRACT

INTRODUCTION: Cherubism is a human genetic disorder that causes bilateral symmetrical enlargement of the maxilla and the mandible in children. It is caused by mutations in SH3BP2. The exact pathogenesis of the disorder is an area of active research. Sh3bp2 knock-in mice were developed by introducing a Pro416Arg mutation (Pro418Arg in humans) in the mouse genome. The osteoclast phenotype of this mouse model was recently described. METHODS: We examined the bone phenotype of the cherubism mouse model, the role of Sh3bp2 during bone formation, osteoblast differentiation, and osteoblast function. RESULTS: We observed delays in early postnatal development of homozygous Sh3bp2(KI/KI) mice, which exhibited increased growth plate thickness and significantly decreased trabecular bone thickness and bone mineral density. Histomorphometric and microcomputed tomography analyses showed bone loss in the cranial and appendicular skeletons. Sh3bp2(KI/KI) mice also exhibited a significant decrease in osteoid formation that indicated a defect in osteoblast function. Calvarial osteoblast cell cultures had decreased alkaline phosphatase expression and mineralization, suggesting reduced differentiation potential. Gene expression of osteoblast differentiation markers such as collagen type I, alkaline phosphatase, and osteocalcin were decreased in osteoblast cultures from Sh3bp2(KI/KI) mice. CONCLUSIONS: These data suggest that Sh3bp2 regulates bone homeostasis through not only osteoclast-specific effects, but also through effects on osteoblast differentiation and function.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cherubism/genetics , Osteoblasts/physiology , Osteogenesis/physiology , Adaptor Proteins, Signal Transducing/genetics , Alkaline Phosphatase/metabolism , Amino Acid Substitution , Animals , Bone Density/genetics , Bone Density/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Cherubism/metabolism , Collagen Type I/metabolism , Disease Models, Animal , Female , Gene Expression Regulation/physiology , Gene Knock-In Techniques , Male , Mice , Mice, Mutant Strains , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteogenesis/genetics
8.
Mol Cell Biol ; 30(1): 43-53, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19884350

ABSTRACT

Nascent-polypeptide-associated complex and coactivator alpha (alpha NAC) is a protein shuttling between the nucleus and the cytoplasm. Upon phosphorylation at residue serine 43 by integrin-linked kinase, alpha NAC is translocated to the nuclei of osteoblasts, where it acts as an AP-1 coactivator to increase osteocalcin gene transcription. To determine the physiological role of nuclear alpha NAC, we engineered a knock-in mouse model with a serine-to-alanine mutation at position 43 (S43A). The S43A mutation resulted in a decrease in the amount of nuclear alpha NAC with reduced osteocalcin gene promoter occupancy, leading to a significant decrease in osteocalcin gene transcription. The S43A mutant bones also expressed decreased levels of alpha(1)(I) collagen mRNA and as a consequence had significantly less osteoid tissue. Transient transfection assays and chromatin immunoprecipitation confirmed the alpha(1)(I) collagen gene as a novel alpha NAC target. The reduced quantity of bone matrix in S43A mutant bones was mineralized faster, as demonstrated by the significantly reduced mineralization lag time, producing a lower volume of immature, woven-type bone characterized by poor lamellation and an increase in the number of osteocytes. Accordingly, the expression of the osteocyte differentiation marker genes DMP-1 (dentin matrix protein 1), E11, and SOST (sclerostin) was increased. The accelerated mineralization phenotype was cell autonomous, as osteoblasts isolated from the calvaria of S43A mutant mice mineralized their matrix faster than did wild-type cells. Thus, inhibition of alpha NAC nuclear translocation results in an osteopenic phenotype caused by reduced expression of osteocalcin and type I collagen, accelerated mineralization, and immature woven-bone formation.


Subject(s)
Bone Matrix/physiology , Calcification, Physiologic/physiology , Cell Nucleus/metabolism , Molecular Chaperones/physiology , Osteoblasts/physiology , Active Transport, Cell Nucleus , Animals , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/pathology , Cell Count , Cells, Cultured , Collagen Type I/biosynthesis , Collagen Type I/genetics , Gene Knock-In Techniques , Mice , Mice, Mutant Strains , Molecular Chaperones/genetics , Osteocalcin/biosynthesis , Osteocalcin/genetics , Polymorphism, Single Nucleotide , Transcription Factor AP-1/physiology , Transcription, Genetic
9.
Am J Med Genet A ; 135(3): 274-7, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15887275

ABSTRACT

Abnormalities in folate and/or homocysteine metabolism may adversely influence embryonic development, leading to the birth of infants with a variety of congenital malformations, including neural tube defects (NTDs) and craniofacial abnormalities. Based upon suggestive evidence that periconceptional folic acid supplementation is effective in preventing a significant proportion of the aforementioned birth defects, genetic variation in the folate biosynthetic pathways may influence the infant's susceptibility to these birth defects. The goal of our study was to investigate sequence variations in the betaine-homocysteine methyltransferase (BHMT) and betaine-homocysteine methyltransferase (BHMT2) genes as modifiers of risk of spina bifida, cleft palate, and cleft lip and palate. The results of this study indicated that individuals homozygous for the single nucleotide polymorphism R239Q in BHMT did not have elevated risks for spina bifida. Genotype frequencies for the BHMT2 rs626105 polymorphism also did not reveal any elevated risks for spina bifida, and only a modest, imprecise elevation of risk for orofacial clefts. The results of these experiments suggest that variants of the BHMT/BHMT2 genes in infants do not substantially contribute to the risk of spina bifida or orofacial clefts in our study population.


Subject(s)
Methyltransferases/genetics , Mouth Abnormalities/genetics , Spinal Dysraphism/genetics , Betaine-Homocysteine S-Methyltransferase , Cleft Lip/diagnosis , Cleft Lip/genetics , Cleft Palate/diagnosis , Cleft Palate/genetics , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Gene Frequency , Genetic Testing , Genotype , Humans , Infant , Infant, Newborn , Isoenzymes/genetics , Linkage Disequilibrium , Mouth Abnormalities/diagnosis , Neonatal Screening , Odds Ratio , Polymorphism, Single Nucleotide , Risk Factors , Spinal Dysraphism/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...